Journal of Bone and Mineral Metabolism (2019) 37:9–17 https://doi.org/10.1007/s00774-018-0971-7

INIVITED REVIEW

Regulatory mechanisms of sclerostin expression during bone remodeling

Masanori Koide1 · Yasuhiro Kobayashi1

Received: 12 September 2018 / Accepted: 14 October 2018 / Published online: 24 October 2018 © The Japanese Society for Bone and Mineral Research and Springer Japan KK, part of Springer Nature 2018

Abstract are embedded in bone matrices and are connected to each other to respond to mechanical loading on bone. Recent studies have demonstrated the roles of mechanical loading in bone accrual. Bone responds to mechanical loading by decreasing the expression of sclerostin, an inhibitor of Wnt/β-catenin signals, in osteocytes. This increases bone mass because the activation of Wnt/β-catenin signals in bone microenvironments promotes bone formation and suppresses bone resorption. Thus, in recent years, sclerostin have attracted increasing attention in bone metabolism. However, the regulatory mechanism of sclerostin expression during bone remodeling has not been fully elucidated. In this review, we summarized the regulation of bone formation and resorption by Wnt signals, a Wnt/β-catenin signal inhibitor sclerostin, and molecular mechanisms by which the expression of sclerostin is suppressed by mechanical loading and . We also discuss a possibility that osteoclasts suppress the expression of sclerostin during bone remodeling, which in turn, promote bone formation. The efectiveness of an anti-sclerostin with anti-dickkopf-1 antibody for increasing bone mass was discussed.

Keywords Sclerostin · Bone remodeling · Osteocytes · · Osteoclasts

Introduction osteoclast precursors. (OPG), a decoy receptor of RANKL, is expressed by osteoblasts and osteo- Bone is continuously remodeled throughout life to main- cytes. OPG interferes with the interaction between RANKL tain plasma calcium homeostasis and prevent the accumu- and RANK, which in turn, inhibits osteoclast diferentiation lation of old bone. Osteoclasts resorb bone, and new bone [6, 7]. is then formed by osteoblasts to replace the amount of bone OPG-knockout (KO) mice exhibited increased bone resorbed. This is called bone remodeling, and bone mass resorption due to excess osteoclast diferentiation and activ- is kept constant by balanced bone remodeling due to the ity [8, 9]. In addition, the activity of bone formation mark- coupling between bone resorption and formation [1, 2]. An ers, such as serum alkaline phosphatase, and bone formation imbalance between bone resorption and formation leads to parameters were also increased [8, 10], suggesting that bone either a low bone mass or a high bone mass [1]. formation was also enhanced in OPG-KO mice even though Osteoclasts differentiate from osteoclast precursors, bone mass was decreased. When these mice were treated monocyte–macrophage lineage cells, by stimulation by with the anti-bone-resorbing drug , bone for- receptor activator of NF-κB ligand (RANKL) and mac- mation was markedly suppressed in association with the sup- rophage colony-stimulating factors (M-CSF) [3–5]. RANKL pression of bone resorption [10]. These results demonstrated and M-CSF bind to receptor activator of NF-κB (RANK) that bone resorption is linked to bone formation by factors and c-fms, respectively. Both receptors are expressed in facilitating the transition from bone resorption to formation, the so-called coupling factors. Several factors have been found to act as coupling factors * Yasuhiro Kobayashi [email protected] between bone resorption and formation [1, 2]. Transforming growth factor beta (TGF-β) and insulin-like growth factor 1 Division of Hard Tissue Research, Institute for Oral Science, I (IGF-I) are embedded in bone matrices during bone for- Matsumoto Dental University, 1780 Gobara, Hiro‑oka, mation [11, 12]. These cytokines are released from bone Shiojiri, Nagano 399‑0781, Japan

Vol.:(0123456789)1 3 10 Journal of Bone and Mineral Metabolism (2019) 37:9–17 matrices during bone resorption and they then induce the migration of precursors to bone resorption sites. Osteoclast-derived factors, such as cardiotrophin-1 (CT-1), Wnt10b, sphingosine-1-phosphate, BMP-6, collagen triple- helix repeat-containing 1, and platelet-derived growth fac- tor (PDGF)-BB, were reported to directly act on osteoblast lineage cells, and promote the diferentiation of osteoblasts and bone formation [13–17]. EphrinB2, expressed by oste- oclasts, also promotes the differentiation of osteoblasts through EphB4 receptors [18]. Osteocytes reportedly produce several Wnt ligands including Wnt1 [19] and Wnt/β-catenin signal inhibi- tor sclerostin [20]. The loss-of-function mutations of the SOST (encoding sclerostin) cause osteosclerosis with abnormal high bone mass, suggesting that bone formation is usually suppressed by sclerostin through the inhibition of Wnt/β-catenin signals to achieve normal bone development. Fig. 1 Wnt signals. Wnt ligands activate β-catenin-dependent canoni- However, it is not fully examined whether osteoclast-derived cal and -independent non-canonical signals. Wntless, an 8-pass factors also act on osteocytes to inhibit the expression of transmembrane , is necessary for Wnt secretion. Wntless-def- sclerostin, which in turn, promote bone formation. cient cells failed to secrete all Wnt ligands. Wnt ligands bind to the receptor complex of frizzled and LRP5/6, and inhibit the activity of In this review, we introduced roles of Wnt signals in bone β-catenin destruction complexes. This leads to cytosolic accumulation formation and regulatory mechanism of sclerostin expres- and nuclear translocation of β-catenin. Nuclear β-catenin together sion by PTH. We would like to propose a possibility that with TCF/LEF induces the transcription of the target . Scle- osteoclasts suppress the expression of sclerostin during bone rostin and Dkk-1 bind to LRP5/6, and inhibit Wnt/β-catenin signals. Binding of sclerostin to LRP4 is required for the inhibitory action of remodeling, which in turn, promote bone formation [21]. In sclerostin. Wnt ligands such as Wnt5a binds receptor complex of friz- addition, we discussed the efects of anti-Dkk-1 and anti- zled and Ror1/2, which in turn activates β-catenin-independent non- sclerostin on bone accrual. canonical signals

Regulation of bone formation by Wnt/ also increased in DA-β-catenin Ocy mice, thereby increas- β‑catenin signals ing bone resorption. These fndings suggest that osteocytes as well as osteoblasts play a critical role in bone anabolic Wnt ligands activate β-catenin-dependent canonical and action of Wnt/β-catenin signals although the mechanism -independent non-canonical signaling pathways [22] by which the expression of RANKL was increased in (Fig. 1). Canonical and non-canonical Wnt signals regu- DA-β-catenin Ocy mice remains to be elucidated. late bone formation and resorption [23]. At present, 19 Wnt Non-canonical Wnt signals impact bone resorption. ligands have been identifed in humans and mice [22]. Wnt Non-canonical Wnt5a binds to its receptor Ror2 in osteo- ligands, such as Wnt1, Wnt5a, Wnt7b, and Wnt10b, report- clast precursors and promotes the expression of RANK, edly regulate bone formation [19, 24–27]. which in turn increased RANKL-induced osteoclast Analysis of mice having gain-of-function and loss- formation [24]. Wnt5a-Ror2 signals also promote bone- of-function mutations of β-catenin in mature osteoblasts resorbing activity of osteoclasts through small GTPase revealed that Wnt/β-catenin signals markedly increase Rho-Pkn3-c-Src signaling axis [30]. In contrast to Wnt5a, bone mass due to the decreased bone resorption without Wnt16 reportedly activated canonical Wnt signals in oste- afecting bone formation [28]. This study has shown that oblastic cells and non-canonical signals in osteoclast pre- activation of Wnt/β-catenin signals in mature osteoblasts cursors [31]. Wnt16-defcient mice exhibited low cortical promotes OPG expression, thereby inhibiting osteoclas- but not trabecular bone mass [31]. Osteoblastogenesis is togenesis. Ten years later, mice expressing dominant normal in those Wnt16-defcient calvaria-derived osteo- active form of β-catenin in osteocytes (DA-β-catenin Ocy blasts in cultures. Treatment of osteoblastic MC3T3-E1 mice) reportedly exhibited high bone mass associated with cells with Wnt16 increased the expression of OPG through increased bone resorption and formation [29]. Similar to the Wnt/β-catenin signals. Furthermore, Wnt16 inhibited mice expressing constitutively active form of β-catenin RANKL-induced osteoclast formation by suppression of in mature osteoblasts [28], the expression of OPG was NF-κB and NFATc1 signals [31]. These results suggest increased in those mice. The expression of RANKL was that Wnt16 inhibits RANK signals in osteoclast precursors

1 3 Journal of Bone and Mineral Metabolism (2019) 37:9–17 11 and induces the expression of OPG in osteoblasts, which bone mass [44, 45]. These fndings suggest that sclerostin in turn, inhibits osteoclast formation. inhibits Wnt/β-catenin signals and suppresses bone forma- Wntless (Wls), an 8-pass transmembrane protein, is tion (Fig. 1). required for the secretion of Wnt ligands [32, 33] (Fig. 1). LRP4 is reportedly required for the inhibitory action of Therefore, Wnt ligands cannot be secreted from Wls-def- sclerostin (46). Two mutations in the LRP4 gene (R1170W cient cells. Both canonical and non-canonical Wnt signals and W1186S) have been found in patients with sclerosteo- are inactivated in those cells. As several kinds of Wnt sis [46]. Sclerostin failed to bind these LRP4 mutants and ligands are secreted from osteoblasts, Wls-defcient osteo- inhibit Wnt/β-catenin signals in cells expressing these blasts are useful to analyze the roles of osteoblast-derived mutants. These fndings indicated that LRP4 bound to scle- Wnt ligands in bone formation. Osteoblast-specifc Wls-con- rostin to facilitate the inhibitory action of sclerostin. Osteo- ditional knockout (cKO) mice exhibit a marked decrease in blast-specifc Lrp4 cKO mice (Lrp4fox/fox; osteocalcin-cre trabecular bone and cortical bone [34]. Histomorphometric mice) exhibited a high bone mass with increased bone for- analysis revealed a signifcant reduction of the bone forma- mation [47, 48]. These fndings strongly indicate that LRP4 tion rate in Wls-cKO mice [34]. Furthermore, the expres- facilitates the inhibitory action of sclerostin in vivo. sion of osteoblast marker genes, such as Runx2 and Osterix, Dickkopf-1 (Dkk-1) is also an inhibitor of Wnt/β-catenin as well as the Wnt target gene Axin2 was significantly signals. Dkk-1 binds to LRP5/6 receptors and a transmem- decreased in Wls-defcient osteoblasts [34]. Mineralization brane protein kremen, and the complex of Dkk-1, kremen, was also decreased in these cells. These fndings suggested and Lrp5/6 internalizes from cell surfaces, thereby inhibit- that Wnt ligands secreted from osteoblast lineage cells are ing Wnt/β-catenin signals [49]. Dkk-1 is highly expressed important for osteoblast diferentiation and bone formation. in bone tissue. Mice having heterozygous deletion of the Low-density lipoprotein receptor-related protein (LRP) Dkk-1 gene exhibited increased bone mass [50], whereas 5/6 functions as a co-receptor for Wnt/β-catenin signals. Dkk-1 transgenic mice exhibited decreased bone mass [51]. Analysis of loss-of-function and gain-of-function muta- Bone resorption markers such as urine deoxypyridinoline tions in the Lrp5 gene demonstrated that Wnt/β-catenin sig- and the number of osteoclasts remained unchanged in those nals promote osteoblast diferentiation and increase bone mice, suggesting that Dkk-1 is not strongly involved in mass [35]. Furthermore, Osterix failed to be expressed by osteoclast formation during developmental process [50]. β-catenin-defcient perichondrial cells or periosteal cells [36, Furthermore, administration of an anti-Dkk-1 neutralizing 37]. Thus, Wnt/β-catenin signals are necessary for osteoblast antibody reportedly increased bone mass by promoting bone diferentiation and bone formation. formation [52]. These fndings demonstrate that Wnt/β- catenin signals in bone are tightly regulated by Dkk-1 and bone-specifc sclerostin. Roles of Wnt inhibitors in bone formation

The SOST gene is a responsible gene for osteosclero- Efects of mechanical loading sis and van Buchem disease. A loss-of-function mutation on the sclerostin expression in the SOST gene causes sclerosteosis with an abnormal increase in bone mass [38, 39]. A homozygous 52-kb non- Osteocytes sense changes in bone fluid flow caused by coding deletion (ECR5 region) of the SOST gene causes Van mechanical loading, and are involved in the increase and Buchem disease, which presents the same clinical features maintenance of bone mass [53]. Loading in upper limbs as sclerosteosis [40]. The ECR5 region in the SOST gene is reduced the expression of sclerostin and Dkk-1 in ulna- a bone-specifc enhancer of sclerostin expression in osteo- loading model mice [54]. In contrast, unloading increased cytes [41]. Sclerostin, a glycoprotein secreted from osteo- the expression of sclerostin in the lower limbs of tail-sus- cytes, binds to the frst two YWTD-EGF repeat domains pended mice and then decreased bone formation [55]. Bone of LRP5/6 and inhibits Wnt/β-catenin signals [42]. Similar formation failed to decrease in the unloaded lower limbs of to sclerosteosis, Sost-KO mice exhibited high bone mass Sost-KO mice [55]. Furthermore, transgenic mice express- with a marked increase in bone formation [43]. Bone resorp- ing human sclerostin in osteocytes exhibited the suppres- tion parameters such as serum TRAP5b and the number of sion of mechanical loading-induced bone formation [56]. osteoclasts remained unchanged, indicating that the def- These fndings revealed that mechanical loading suppresses ciency of sclerostin does not afect osteoclast formation. the expression of sclerostin in osteocytes, which in turn, This fnding suggests that sclerostin is not strongly involved promotes bone formation. in osteoclast formation during developmental process. Fur- Periostin is a secreted extracellular matrix protein thermore, administration of anti-sclerostin neutralizing anti- expressed in the periosteum and periodontal ligaments [57]. body reportedly promoted bone formation and increased Periostin (encoded by the Postn gene)-KO mice developed

1 3 12 Journal of Bone and Mineral Metabolism (2019) 37:9–17 osteoporosis and periodontitis [58]. Mechanical loading also reportedly induced the expression of periostin and sup- pressed sclerostin expression [59]. Bonnet et al. [59, 60] ana- lyzed the expression of sclerostin in osteocytes and the bone mass of cortical bone in Postn-KO mice treated with PTH or mechanical loading. In Postn-KO mice, administration of PTH or mechanical loading failed to decrease the expression of sclerostin, and the bone mass in cortical bone was not increased. In contrast, administration of an anti-sclerostin neutralizing antibody increased the bone mass in cortical bone of Postn-KO mice. Therefore, administration of PTH or mechanical loading increased the expression of periostin in the periosteum, and the increased periostin reduced the expression of sclerostin, which in turn, promoted bone for- mation in cortical bone (Fig. 2). These results suggest a new function of periostin as a suppressor of sclerostin expression. Fig. 3 PTH signals suppress the expression of sclerostin. MEF2 binds The mechanisms by which periostin suppresses the expres- the ECR5 (enhancer region) of the Sost gene to induce the expression sion of sclerostin need to be clarifed in the future. of sclerostin. Salt-induced kinases (SIKs) phosphorylate HDAC4/5 to promote complex formation of HDAC4/5 and 14-3-3, which in turn retains HDAC4/5 in the cytoplasm. PTH signals inhibit the kinase activity of SIKs, which in turn increases dephosphorylated HDAC Regulatory mechanism of sclerostin in nucleus. Nuclear dephosphorylated HDAC inhibits the activity of expression by PTH and IL‑6 family cytokines MEF2

As described above, the down-regulation of sclerostin of MEF2 to the ECR5 region and decreased the expression expression is considered to be an important step for bone of sclerostin [65]. accrual. Because the efect of PTH on the expression of scle- The detailed roles of MEF2 in the expression of scle- rostin is well-studied, here we would like to introduce the rostin have been proposed. Histone deacetylase (HDAC) 5 inhibitory action of PTH in sclerostin expression. Intermit- binds to MEF2 and suppresses the expression of sclerostin tent administration of PTH increases bone mass [61, 62]. [66] (Fig. 3). The expression of sclerostin was enhanced As PTH markedly suppresses the expression of sclerostin in HDAC5-KO mice, which exhibited decreased bone in osteocytes, the role of PTH in bone accrual is consid- formation [66], suggesting that HDAC5 suppresses the ered to be mediated, in part, by the inhibition of sclerostin expression of sclerostin. Knockdown of HDAC5 enhanced expression [63, 64]. The 52-kb region (ECR5) containing the transcriptional activity of MEF2 and the expression the enhancer downstream of the Sost gene is important for of sclerostin. Taken together, the association of HDAC5 bone-specifc sclerostin expression [40] (Fig. 3). Myocyte with MEF2 in osteocytes reduced the transcription activ- enhancer factor (MEF) 2, a transcription factor, binds to the ity of MEF2, and subsequently the expression of sclerostin. ECR5 region and induces the expression of sclerostin [65]. Recently, a further detailed mechanism was reported. Salt- Stimulation of PTH in osteocytes reduced the recruitment inducible kinases (SIKs), which are a kinase for HDAC4/5 and cAMP-regulated transcriptional coactivators (CRTC) [67], were found to phosphorylate HDAC5 to promote the complex formation of HDAC5 with 14-3-3 . 14-3-3 proteins are intracellular dimeric proteins involved in vari- ous biological events such as signal transductions. 14-3-3 retained HDAC5 in the cytoplasm [68, 69]. Stimulation of PTH suppressed the activity of SIKs and increased dephos- phorylated HDAC4/5, which localizes to the nucleus. The nuclear HDAC4/5 forms a complex with MEF2 and inhibits the transcriptional activity of MEF2, which in turn, sup- presses the expression of sclerostin [69] (Fig. 3). Further- more, SIK inhibitors have been reported to promote bone formation. Of note, the suppression of SIK activity was Fig. 2 Roles of periostin in the suppression of sclerostin expression. Periostin secreted from periosteal osteoblasts acts on osteocytes and demonstrated to increase the expression of RANKL via inhibits the expression of sclerostin nuclear translocation of dephosphorylated CRTC 2 [69].

1 3 Journal of Bone and Mineral Metabolism (2019) 37:9–17 13

This suggests that SIK is also involved in osteoclastogenesis suppresses the expression of sclerostin and when osteoblast linage cells are stimulated by PTH. In the increases that of RANKL to induce osteoclast formation future, clarifcation as to whether SIKs and HDAC4/5 are during bone remodeling. They also suggest that osteoclast- involved in the mechanical loading-induced suppression of derived CT-1 also suppresses the expression of sclerostin. sclerostin expression is needed. However, it is unknown how CT-1 regulates the expression Roles of PTH1 receptor (PTH1R) in osteocytes in the of sclerostin in vivo. expression of sclerostin have been examined using dentin matrix protein (DMP)-1-Cre: PTH1R conditional knock- out mice. Powell et al. [70] prepared tamoxifen-inducible Roles of sclerostin in bone remodeling PTH1R cKO mice using DMP-1-CreERT2 mice. These PTH1R cKO mice exhibited low bone mass with the During bone remodeling, several coupling factors are con- increased expression of sclerostin mRNA. The expression sidered to link bone resorption with formation because of Wnt target gene axin was decreased in bone tissues from bone resorption promotes bone formation [1, 2]. Excellent those mice. These results suggest that PTH1R signals sup- review articles describe roles of osteocytes in bone remod- press the expression of sclerostin, which in turn, promotes eling [73–75]. Osteocytes response to mechanical loading bone accrual under physiological condition. In contrast to and PTH, and then decrease the expression of sclerostin, tamoxifen-inducible PTH1R cKO mice [70], PTH1R cKO thereby promoting bone formation. Osteocytes also produce mice using non-inducible DMP-1 Cre exhibited high bone RANKL to induce bone resorption. Thus, osteocytes regu- mass with a modest decrease in bone resorption. These late bone remodeling. results suggest that PTH1R signal in osteocytes may not As mentioned above, OPG-KO mice exhibited high bone be involved in the suppression of sclerostin expression by turnover with increased bone resorption and formation. The PTH [71]. However, the expression of Wnt target genes such expression of sclerostin was reportedly lower in OPG-KO as Naked2 and Conexin 43 were decreased in those mice, mice [76]. However, the mechanisms have not been fully suggesting that Wnt/β-catenin signals suppressed probably elucidated at the molecular level. We analyzed OPG-KO due to the increase in the expression of sclerostin. Further- mice and found that osteoclasts promoted bone formation more, the anabolic action and the suppression of sclerostin via the suppression of sclerostin expression in osteocytes expression by intermittent injections of PTH disappeared in [21]. Antibody array analysis and real-time PCR analysis those cKO mice [71], suggesting that PTH1R signals play demonstrated that mature osteoclasts abundantly expressed an important role in anabolic action of PTH by the sup- and secreted LIF [21]. LIF secreted from osteoclasts pression of sclerostin expression. Those cKO mice exhibited reduced the expression of sclerostin in osteocytes and cul- the decreased expression of RANKL and the decrease in tured UMR106 cells [21]. Anti-LIF antibody, but not anti- bone resorption marker serum CTX. These fndings suggest oncostatin M antibody, neutralized the suppressive efects of that bone mass is increased due to the suppression of bone osteoclast-conditioned medium on sclerostin expression in resorption in those cKO mice. The increased bone mass in UMR106 cells [21], suggesting that osteoclast-conditioned those cKO mice might be due to cre recombinase activity medium contains enough LIF to suppress the expression of during developmental process. Further studies are needed sclerostin. Furthermore, administration of the anti-RANKL to clarify this point. antibody inhibited osteoclast diferentiation and inhibited Recently, we found that osteoclast-derived leukemia bone resorption in OPG-KO mice, and increased the expres- inhibitory factor (LIF) suppresses the expression of scle- sion of sclerostin by osteocytes [21]. Wnt/β-catenin signals rostin in UMR106 cells [21]. Therefore, we introduce efects in bone tissues were decreased in mice treated with the anti- of IL-6 family cytokines such as LIF, oncostatin M, and RANKL antibody, which in turn, suppressed bone forma- CT-1 on the expression of sclerostin. These IL-6 family tion [21]. These results suggest that regulation of sclerostin members reportedly suppress the expression of sclerostin expression by osteoclasts plays an important role in the in cultured UMR106 cells [72]. Oncostatin M was reported coupling between bone resorption and formation (Fig. 4). to be abundantly expressed in osteoblasts [72]. Furthermore, A clinical study found that the administration of the anti- administration of recombinant oncostatin M decreased the RANKL antibody reduced bone formation markers associ- expression of sclerostin in wild-type mice [72]. IL-6 family ated with the suppression of bone resorption markers [77, cytokines such as IL-6 and oncostatin M reportedly induced 78]. Moreover, administration of the anti-RANKL antibody the expression of RANKL to promote osteoclast formation increased the amount of sclerostin in serum [79, 80]. These in co-cultures of bone marrow cells with osteoblastic cells clinical studies strongly suggested that bone resorption sup- [5]. The treatment with oncostatin M also increased the presses the expression of sclerostin, which in turn, promotes expression of RANKL in osteoblastic cell line KUSA 4b10 bone formation in humans. Thus, simultaneous administra- cells [72]. These results indicate that osteoblast-derived tion of anti-sclerostin antibodies with RANKL antibodies

1 3 14 Journal of Bone and Mineral Metabolism (2019) 37:9–17

of sclerostin. In contrast, treatment of ovariectomized rats with the anti-sclerostin antibody increased the expression of Dkk-1[83]. Administration of both anti-Dkk-1 and anti-scle- rostin antibodies more markedly increased the bone volume than only Dkk-1 antibody or sclerostin antibody in fracture model rats [83]. In addition, administration of a bispecifc antibody targeting sclerostin and Dkk-1 promoted fracture healing as compared with the anti-sclerostin antibody [84]. These results suggest that the inhibition of both sclerostin and Dkk-1 is useful for promoting bone formation compared with neutralizing either sclerostin or Dkk-1, and that the inhibition of sclerostin induces the expression of Dkk-1 Fig. 4 LIF secreted from osteoclasts inhibits the expression of scle- because Dkk-1 is a target gene of Wnt/β-catenin signals. rostin in osteocytes. Osteocytes produce sclerostin, which inhib- its Wnt/β-catenin signals in osteoblasts and suppresses excess bone formation. Osteoclasts secrete LIF (1) and inhibit the expression of sclerostin (2), which in turn, promotes bone formation through the activation of Wnt/β-catenin signals (3). Osteocytes are involved in the Conclusion coupling between bone resorption and formation In this review, we introduced the regulatory mechanism of maybe efective for keeping bone formation rate, because sclerostin expression, roles of osteocytes in the coupling administration of anti-RANKL antibodies suppresses bone between bone resorption and formation, and the useful- formation due to up-regulation of sclerostin expression. It is ness of sclerostin and Dkk-1 inhibition for bone accrual also important that the regulatory mechanisms of sclerostin and fracture treatment. Osteoclasts secrete LIF and inhibit expression by bone resorption be elucidated in more detail the expression of sclerostin, which in turn, promote bone in the future. formation through the activation of Wnt/β-catenin signals. Although recent studies established important roles of scle- rostin in bone metabolism, several molecular mechanisms, Anti‑sclerostin antibody and anti‑DKK1 such as the suppression of sclerostin expression by mechani- antibody cal loading and LIF, remain to be elucidated. These issues will be solved in future studies, leading to the development Recently, Witcher et al. [81] reported that conditional dele- of drugs for osteoporosis. tion of the Dkk-1 gene using DMP-1 Cre and treatment of Acknowledgements This work was supported in part by Grants-in-Aid wild-type mice with an anti-Dkk-1 antibody had negligible 18H02980 (M.K.), 16H02691 (Y. K.), 18H05388 (Y. K.) from the Min- efects on bone accrual. Administration of the anti-Dkk-1 istry of Education, Culture, Sports, Science and Technology of Japan. antibody increased the expression of sclerostin [81], sug- gesting that it is regulated by Wnt/β-catenin signals. This Compliance with ethical standards fnding is consistent with the previous fnding that genetic activation of β-catenin in osteocytes increases the expression Conflict of interest The authors declare that they have no confict of of sclerostin in bone [82]. Thus, Wnt/β-catenin signals posi- interest. tively regulate the expression of sclerostin. Further studies are needed to clarify whether Wnt/β-catenin signals regulate the transcription activity of the Sost gene. Administration of References the anti-Dkk-1 antibody to Sost-KO mice further enhanced bone formation even though Sost-KO mice exhibited high 1. Sims NA, Martin TJ (2014) Coupling the activities of bone for- bone mass. The combined administration of anti-DKK-1 and mation and resorption: a multitude of signals within the basic anti- sclerostin antibodies markedly promoted bone forma- multicellular unit. Bonekey Rep 3:481 tion. These results suggest that the increased expression of 2. Crane JL, Cao X (2014) Bone marrow mesenchymal stem cells and TGF-beta signaling in bone remodeling. J Clin Invest sclerostin by inhibition of Dkk-1 weakens the stimulatory 124:466–472 efects of the anti-Dkk-1 antibody on bone formation. In 3. Yasuda H, Shima N, Nakagawa N, Yamaguchi K, Kinosaki M, addition, postnatal global deletion of the Dkk-1 gene report- Mochizuki S, Tomoyasu A, Yano K, Goto M, Murakami A, Tsuda edly caused high bone mass [83]. Of note, the expression E, Morinaga T, Higashio K, Udagawa N, Takahashi N, Suda T (1998) Osteoclast diferentiation factor is a ligand for osteopro- of sclerostin was higher in these mice [83]. This also sug- tegerin/osteoclastogenesis-inhibitory factor and is identical to gests that Wnt/β-catenin signals promote the expression TRANCE/RANKL. Proc Natl Acad Sci USA 95:3597–3602

1 3 Journal of Bone and Mineral Metabolism (2019) 37:9–17 15

4. Kong YY, Feige U, Sarosi I, Bolon B, Tafuri A et al (1999) Acti- osteoblast function during bone homeostasis. J Clin Invest vated T cells regulate bone loss and joint destruction in adjuvant 127:2678–2688 arthritis through osteoprotegerin ligand. Nature 402:304–309 20. van Bezooijen RL, Roelen BA, Visser A, van der Wee-Pals L, de 5. Suda T, Takahashi N, Udagawa N, Jimi E, Gillespie MT, Martin Wilt E, Karperien M, Hamersma H, Papapoulos SE, ten Dijke P, TJ (1999) Modulation of osteoclast diferentiation and function by Löwik CW (2004) Sclerostin is an -expressed negative the new members of the tumor necrosis factor receptor and ligand regulator of bone formation, but not a classical BMP antagonist. families. Endocr Rev 20:345–357 J Exp Med 199:805–814 6. Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS et al 21. Koide M, Kobayashi Y, Yamashita T, Uehara S, Nakamura M, (1997) Osteoprotegerin: a novel secreted protein involved in the Hiraoka BY, Ozaki Y, Iimura T, Yasuda H, Takahashi N, Udagawa regulation of bone density. Cell 89:309–319 N (2017) Bone Formation is coupled to resorption via suppres- 7. Yasuda H, Shima N, Nakagawa N, Mochizuki SI, Yano K, Fujise sion of sclerostin expression by osteoclasts. J Bone Miner Res N, Sato Y, Goto M, Yamaguchi K, Kuriyama M, Kanno T, 32:2074–2086 Murakami A, Tsuda E, Morinaga T, Higashio K (1998) Identity 22. Miller JR (2002) The Wnts. Genome Biol 3:Reviews3001 of osteoclastogenesis inhibitory factor (OCIF) and osteoprotegerin 23. Baron R, Kneissel M (2013) WNT signaling in bone homeosta- (OPG): a mechanism by which OPG/OCIF inhibits osteoclas- sis and disease: from human mutations to treatments. Nat Med togenesis in vitro. Endocrinology 139:1329–1337 19:179–192 8. Bucay N, Sarosi I, Dunstan CR, Morony S, Tarpley J, Capparelli 24. Maeda K, Kobayashi Y, Udagawa N, Uehara S, Ishihara A, Mizo- C, Scully S, Tan HL, Xu W, Lacey DL, Boyle WJ, Simonet WS guchi T, Kikuchi Y, Takada I, Kato S, Kani S, Nishita M, Marumo (1998) osteoprotegerin-defcient mice develop early onset osteo- K, Martin TJ, Minami Y, Takahashi N (2012) Wnt5a-Ror2 sign- porosis and arterial calcifcation. Genes Dev 12:1260–1268 aling between osteoblast-lineage cells and osteoclast precursors 9. Mizuno A, Amizuka N, Irie K, Murakami A, Fujise N, Kanno enhances osteoclastogenesis. Nat Med 18:405–412 T, Sato Y, Nakagawa N, Yasuda H, Mochizuki S, Gomibuchi T, 25. Okamoto M, Udagawa N, Uehara S, Maeda K, Yamashita T, Yano K, Shima N, Washida N, Tsuda E, Morinaga T, Higashio K, Nakamichi Y, Kato H, Saito N, Minami Y, Takahashi N, Kob- Ozawa H (1998) Severe osteoporosis in mice lacking osteoclas- ayashi Y (2014) Noncanonical Wnt5a enhances Wnt/beta-catenin togenesis inhibitory factor/osteoprotegerin. Biochem Biophys Res signaling during osteoblastogenesis. Sci Rep 4:4493 Commun 247:610–615 26. Chen J, Tu X, Esen E, Joeng KS, Lin C, Arbeit JM, Ruegg MA, 10. Nakamura M, Udagawa N, Matsuura S, Mogi M, Nakamura H, Hall MN, Ma L, Long F (2014) WNT7B promotes bone formation Horiuchi H, Saito N, Hiraoka BY, Kobayashi Y, Takaoka K, in part through mTORC1. PLoS Genet 10:e1004145 Ozawa H, Miyazawa H, Takahashi N (2003) Osteoprotegerin 27. Bennett CN, Longo KA, Wright WS, Suva LJ, Lane TF, Hanken- regulates bone formation through a coupling mechanism with son KD, MacDougald OA (2005) Regulation of osteoblas- bone resorption. Endocrinology 144:5441–5449 togenesis and bone mass by Wnt10b. Proc Natl Acad Sci USA 11. Tang Y, Wu X, Lei W, Pang L, Wan C, Shi Z, Zhao L, Nagy TR, 102:3324–3329 Peng X, Hu J, Feng X, Van Hul W, Wan M, Cao X (2009) TGF- 28. Glass DA 2nd, Bialek P, Ahn JD, Starbuck M, Patel MS, Clev- beta1-induced migration of bone mesenchymal stem cells couples ers H, Taketo MM, Long F, McMahon AP, Lang RA, Karsenty bone resorption with formation. Nat Med 15:757–765 G (2005) Canonical Wnt signaling in diferentiated osteoblasts 12. Xian L, Wu X, Pang L, Lou M, Rosen CJ, Qiu T, Crane J, Frassica controls osteoclast diferentiation. Dev Cell 8:751–764 F, Zhang L, Rodriguez JP, Xiaofeng J, Shoshana Y, Shouhong 29. Tu X, Delgado-Calle J, Condon KW, Maycas M, Zhang H, Car- X, Argiris E, Mei W, Xu C (2012) Matrix IGF-1 maintains bone lesso N, Taketo MM, Burr DB, Plotkin LI, Bellido T (2015) Oste- mass by activation of mTOR in mesenchymal stem cells. Nat Med ocytes mediate the anabolic actions of canonical Wnt/β-catenin 18:1095–1101 signaling in bone. Proc Natl Acad Sci USA 112:E478–E486 13. Walker EC, McGregor NE, Poulton IJ, Pompolo S, Allan EH, 30. Uehara S, Udagawa N, Mukai H, Ishihara A, Maeda K, Yamash- Quinn JM, Gillespie MT, Martin TJ, Sims NA (2008) Car- ita T, Murakami K, Nishita M, Nakamura T, Kato S, Minami Y, diotrophin-1 is an osteoclast-derived stimulus of bone forma- Takahashi N, Kobayashi Y (2017) Protein kinase N3 promotes tion required for normal bone remodeling. J Bone Miner Res bone resorption by osteoclasts in response to Wnt5a-Ror2 signal- 23:2025–2032 ing. Sci Signal 10:eaan0023 14. Pederson L, Ruan M, Westendorf JJ, Khosla S, Oursler MJ (2008) 31. Movérare-Skrtic S, Henning P, Liu X, Nagano K, Saito H et al Regulation of bone formation by osteoclasts involves Wnt/BMP (2014) Osteoblast-derived WNT16 represses osteoclastogen- signaling and the chemokine sphingosine-1-phosphate. Proc Natl esis and prevents cortical bone fragility fractures. Nat Med Acad Sci USA 105:20764–20769 20:1279–1288 15. Takeshita S, Fumoto T, Matsuoka K, Park KA, Aburatani H, 32. Bartscherer K, Pelte N, Ingelfnger D, Boutros M (2006) Secretion Kato S, Ito M, Ikeda K (2013) Osteoclast-secreted CTHRC1 of Wnt ligands requires Evi, a conserved transmembrane protein. in the coupling of bone resorption to formation. J Clin Invest Cell 125:523–533 123:3914–3924 33. Banziger C, Soldini D, Schutt C, Zipperlen P, Hausmann G, Basler 16. Lotinun S, Kiviranta R, Matsubara T, Alzate JA, Nef L, Luth A, K (2006) Wntless, a conserved membrane protein dedicated to the Koskivirta I, Kleuser B, Vacher J, Vuorio E, Horne WC, Baron R secretion of Wnt proteins from signaling cells. Cell 125:509–522 (2013) Osteoclast-specifc cathepsin K deletion stimulates S1P- 34. Zhong Z, Zylstra-Diegel CR, Schumacher CA, Baker JJ, Carpen- dependent bone formation. J Clin Invest 123:666–681 ter AC, Rao S, Yao W, Guan M, Helms JA, Lane NE, Lang RA, 17. Xie H, Cui Z, Wang L, Xia Z, Hu Y et al (2014) PDGF-BB Williams BO (2012) Wntless functions in mature osteoblasts to secreted by preosteoclasts induces angiogenesis during coupling regulate bone mass. Proc Natl Acad Sci USA 109:E2197–E2204 with osteogenesis. Nat Med 20:1270–1278 35. Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman S et al 18. Zhao C, Irie N, Takada Y, Shimoda K, Miyamoto T, Nishiwaki T, (2001) LDL receptor-related protein 5 (LRP5) afects bone accrual Suda T, Matsuo K (2006) Bidirectional ephrinB2-EphB4 signal- and eye development. Cell 107:513–523 ing controls bone homeostasis. Cell Metab 4:111–121 36. Hill TP, Spater D, Taketo MM, Birchmeier W, Hartmann C (2005) 19. Joeng KS, Lee YC, Lim J, Chen Y, Jiang MM, Munivez E, Canonical Wnt/beta-catenin signaling prevents osteoblasts from Ambrose C, Lee BH (2017) Osteocyte-specifc WNT1 regulates diferentiating into chondrocytes. Dev Cell 8:727–738

1 3 16 Journal of Bone and Mineral Metabolism (2019) 37:9–17

37. Day TF, Guo X, Garrett-Beal L, Yang Y (2005) Wnt/beta-catenin 52. Glantschnig H, Hampton RA, Lu P, Zhao JZ, Vitelli S et al signaling in mesenchymal progenitors controls osteoblast and (2010) Generation and selection of novel fully human mono- chondrocyte diferentiation during vertebrate skeletogenesis. Dev clonal antibodies that neutralize Dickkopf-1 (DKK1) inhibitory Cell 8:739–750 function in vitro and increase bone mass in vivo. J Biol Chem 38. Brunkow ME, Gardner JC, Van Ness J, Paeper BW, Kovacevich 285:40135–40147 BR, Proll S, Skonier JE, Zhao L, Sabo PJ, Fu Y, Alisch RS, Gillett 53. Bonewald LF, Johnson ML (2008) Osteocytes, mechanosensing L, Colbert T, Tacconi P, Galas D, Hamersma H, Beighton P, Mul- and Wnt signaling. Bone 42:606–615 ligan J (2001) Bone dysplasia sclerosteosis results from loss of 54. Robling AG, Niziolek PJ, Baldridge LA, Condon KW, Allen the SOST gene product, a novel -containing protein. MR, Alam I, Mantila SM, Gluhak-Heinrich J, Bellido TM, Har- Am J Hum Genet 68:577–589 ris SE, Turner CH (2008) Mechanical stimulation of bone in vivo 39. Balemans W, Ebeling M, Patel N, Van Hul E, Olson P et al (2001) reduces osteocyte expression of Sost/sclerostin. J Biol Chem Increased bone density in sclerosteosis is due to the defciency of 283:5866–5875 a novel secreted protein (SOST). Hum Mol Genet 10:537–543 55. Lin C, Jiang X, Dai Z, Guo X, Weng T, Wang J, Li Y, Feng G, Gao 40. Balemans W, Patel N, Ebeling M, Van Hul E, Wuyts W, Lacza X, He L (2009) Sclerostin mediates bone response to mechani- C, Dioszegi M, Dikkers FG, Hildering P, Willems PJ, Verheij cal unloading through antagonizing Wnt/beta-catenin signaling. JB, Lindpaintner K, Vickery B, Foernzler D, Van Hul W (2002) J Bone Miner Res 24:1651–1661 Identifcation of a 52 kb deletion downstream of the SOST gene 56. Tu X, Rhee Y, Condon KW, Bivi N, Allen MR, Dwyer D, Sto- in patients with van Buchem disease. J Med Genet 39:91–97 lina M, Turner CH, Robling AG, Plotkin LI, Bellido T (2011) 41. Collette NM, Genetos DC, Economides AN, Xie L, Shahnazari M, Sost downregulation and local Wnt signaling are required for the Yao W, Lane NE, Harland RM, Loots GG (2012) Targeted dele- osteogenic response to mechanical loading. Bone 50:209–217 tion of Sost distal enhancer increases bone formation and bone 57. Horiuchi K, Amizuka N, Takeshita S, Takamatsu H, Katsuura M, mass. Proc Natl Acad Sci USA 109:14092–14097 Ozawa H, Toyama Y, Bonewald LF, Kudo A (1999) Identifcation 42. Li X, Zhang Y, Kang H, Liu W, Liu P, Zhang J, Harris SE, Wu D and characterization of a novel protein, periostin, with restricted (2005) Sclerostin binds to LRP5/6 and antagonizes canonical Wnt expression to periosteum and periodontal ligament and increased signaling. J Biol Chem 280:19883–19887 expression by transforming growth factor beta. J Bone Miner Res 43. Li X, Ominsky MS, Niu QT, Sun N, Daugherty B, D’Agostin 14:1239–1249 D, Kurahara C, Gao Y, Cao J, Gong J, Asuncion F, Barrero M, 58. Rios H, Koushik SV, Wang H, Wang J, Zhou HM, Lindsley A, Warmington K, Dwyer D, Stolina M, Morony S, Sarosi I, Kosten- Rogers R, Chen Z, Maeda M, Kruzynska-Frejtag A, Feng JQ, uik PJ, Lacey DL, Simonet WS, Ke HZ, Paszty C (2008) Targeted Conway SJ (2005) Periostin null mice exhibit dwarfsm, incisor deletion of the sclerostin gene in mice results in increased bone enamel defects, and an early-onset periodontal disease-like phe- formation and bone strength. J Bone Miner Res 23:860–869 notype. Mol Cell Biol 25:11131–11144 44. Li X, Ominsky MS, Warmington KS, Morony S, Gong J et al 59. Bonnet N, Standley KN, Bianchi EN, Stadelmann V, Foti M, Con- (2009) Sclerostin antibody treatment increases bone formation, way SJ, Ferrari SL (2009) The matricellular protein periostin is bone mass, and bone strength in a rat model of postmenopausal required for sost inhibition and the anabolic response to mechani- osteoporosis. J Bone Miner Res 24:578–588 cal loading and physical activity. J Biol Chem 284:35939–35950 45. Cosman F, Crittenden DB, Adachi JD, Binkley N, Czerwinski 60. Bonnet N, Conway SJ, Ferrari SL (2012) Regulation of beta E, Ferrari S, Hofbauer LC, Lau E, Lewiecki EM, Miyauchi A, catenin signaling and parathyroid hormone anabolic efects in Zerbini CA, Milmont CE, Chen L, Maddox J, Meisner PD, Liba- bone by the matricellular protein periostin. Proc Natl Acad Sci nati C, Grauer A (2016) Romosozumab treatment in postmeno- USA 109:15048–15053 pausal women with osteoporosis. N Engl J Med 375:1532–1543 61. Dempster DW, Cosman F, Parisien M, Shen V, Lindsay R (1993) 46. Leupin O, Piters E, Halleux C, Hu S, Kramer I et al (2011) Bone Anabolic actions of parathyroid hormone on bone. Endocr Rev overgrowth-associated mutations in the LRP4 gene impair scle- 14:690–709 rostin facilitator function. J Biol Chem 286:19489–19500 62. Neer RM, Arnaud CD, Zanchetta JR, Prince R, Gaich GA, Regin- 47. Chang MK, Kramer I, Huber T, Kinzel B, Guth-Gundel S, Leupin ster JY, Hodsman AB, Eriksen EF, Ish-Shalom S, Genant HK, O, Kneissel M (2014) Disruption of Lrp4 function by genetic dele- Wang O, Mitlak BH (2001) Efect of parathyroid hormone (1-34) tion or pharmacological blockade increases bone mass and serum on fractures and bone mineral density in postmenopausal women sclerostin levels. Proc Natl Acad Sci USA 111:E5187–E5195 with osteoporosis. N Engl J Med 344:1434–1441 48. Xiong L, Jung JU, Wu H, Xia WF, Pan JX, Shen C, Mei L, Xiong 63. Keller H, Kneissel M (2005) SOST is a target gene for PTH in WC (2015) Lrp4 in osteoblasts suppresses bone formation and bone. Bone 37:148–158 promotes osteoclastogenesis and bone resorption. Proc Natl Acad 64. Kramer I, Loots GG, Studer A, Keller H, Kneissel M (2010) Para- Sci USA 112:3487–3492 thyroid hormone (PTH)-induced bone gain is blunted in SOST 49. Mao B, Wu W, Davidson G, Marhold J, Li M, Mechler BM, overexpressing and defcient mice. J Bone Miner Res 25:178–189 Delius H, Hoppe D, Stannek P, Walter C, Glinka A, Niehrs C 65. Leupin O, Kramer I, Collette NM, Loots GG, Natt F, Kneissel M, (2002) Kremen proteins are Dickkopf receptors that regulate Wnt/ Keller H (2007) Control of the SOST bone enhancer by PTH using beta-catenin signalling. Nature 417:664–667 MEF2 transcription factors. J Bone Miner Res 22:1957 50. Morvan F, Boulukos K, Clement-Lacroix P, Roman Roman S, 66. Wein MN, Spatz J, Nishimori S, Doench J, Root D, Babij P, Suc-Royer I, Vayssiere B, Ammann P, Martin P, Pinho S, Pog- Nagano K, Baron R, Brooks D, Bouxsein M, Pajevic PD, Kro- nonec P, Mollat P, Niehrs C, Baron R, Rawadi G (2006) Deletion nenberg HM (2015) HDAC5 controls MEF2C-driven sclerostin of a single allele of the Dkk1 gene leads to an increase in bone expression in osteocytes. J Bone Miner Res 30:400–411 formation and bone mass. J Bone Miner Res 21:934–945 67. Berdeaux R, Goebel N, Banaszynski L, Takemori H, Wandless T, 51. Guo J, Liu M, Yang D, Bouxsein ML, Saito H, Galvin RJ, Kuh- Shelton GD, Montminy M (2007) SIK1 is a class II HDAC kinase stoss SA, Thomas CC, Schipani E, Baron R, Bringhurst FR, that promotes survival of skeletal myocytes. Nat Med 13:597–603 Kronenberg HM (2010) Suppression of Wnt signaling by Dkk1 68. Parra M, Verdin E (2010) Regulatory signal transduction path- attenuates PTH-mediated stromal cell response and new bone for- ways for class IIa histone deacetylases. Curr Opin Pharmacol mation. Cell Metab 11:161–171 10:454–460

1 3 Journal of Bone and Mineral Metabolism (2019) 37:9–17 17

69. Wein MN, Liang Y, Goransson O, Sundberg TB, Wang J et al in postmenopausal women with low bone mass: a randomized, (2016) SIKs control osteocyte responses to parathyroid hormone. blinded, phase 3 trial. J Bone Miner Res 24:153–161 Nat Commun 7:13176 78. Reid IR, Miller PD, Brown JP, Kendler DL, Fahrleitner-Pammer 70. Powell WF Jr, Barry KJ, Tulum I, Kobayashi T, Harris SE, Bring- A, Valter I, Maasalu K, Bolognese MA, Woodson G, Bone H, hurst FR, Pajevic PD (2011) Targeted ablation of the PTH/PTHrP Ding B, Wagman RB, San Martin J, Ominsky MS, Dempster DW, receptor in osteocytes impairs bone structure and homeostatic cal- Phase 3 Bone Histology Study G (2010) Efects of cemic responses. J Endocrinol 209:21–32 denosumab on bone histomorphometry: the FREEDOM and 71. Delgado-Calle J, Tu X, Pacheco-Costa R, McAndrews K, Edwards STAND studies. J Bone Miner Res 25:2256–2265 R, Pellegrini GG, Kuhlenschmidt K, Olivos N, Robling A, Pea- 79. Gatti D, Viapiana O, Fracassi E, Idolazzi L, Dartizio C, Povino cock M, Plotkin LI, Bellido T (2017) Control of bone anabolism in MR, Adami S, Rossini M (2012) Sclerostin and DKK1 in post- response to mechanical loading and PTH by distinct mechanisms menopausal osteoporosis treated with denosumab. J Bone Miner downstream of the PTH receptor. J Bone Miner Res 32:522–535 Res 27:2259–2263 72. Walker EC, McGregor NE, Poulton IJ, Solano M, Pompolo S, 80. Anastasilakis AD, Polyzos SA, Gkiomisi A, Bisbinas I, Gerou Fernandes TJ, Constable MJ, Nicholson GC, Zhang JG, Nicola S, Makras P (2013) Comparative efect of versus NA, Gillespie MT, Martin TJ, Sims NA (2010) Oncostatin M pro- denosumab on serum sclerostin and dickkopf-1 levels of naive motes bone formation independently of resorption when signaling postmenopausal women with low bone mass: a randomized, head- through leukemia inhibitory factor receptor in mice. J Clin Invest to-head clinical trial. J Clin Endocrinol Metab 98:3206–3212 120:582–592 81. Witcher PC, Miner SE, Horan DJ, Bullock WA, Lim KE, Kang 73. Bellido T (2014) Osteocyte-driven bone remodeling. Calcif Tissue KS, Adaniya AL, Ross RD, Loots GG, Robling AG (2018) Scle- Int 94:25–34 rostin neutralization unleashes the osteoanabolic efects of Dkk1 74. Goldring SR (2015) The osteocyte: key player in regulating bone inhibition. JCI Insight. https​://doi.org/10.1172/jci.insig​ht.98673​ turnover. RMD Open 1:e000049 82. Tu X, Delgado-Calle J, Condon KW, Maycas M, Zhang H, Car- 75. Prideaux M, Findlay DM, Atkins GJ (2016) Osteocytes: the mas- lesso N, Taketo MM, Burr DB, Plotkin LI, Bellido T (2015) Oste- ter cells in bone remodelling. Curr Opin Pharmacol 28:24–30 ocytes mediate the anabolic actions of canonical Wnt/β-catenin 76. Masuki H, Li M, Hasegawa T, Suzuki R, Ying G, Zhusheng L, signaling in bone. Proc Natl Acad Sci USA 112:E478–E486 Oda K, Yamamoto T, Kawanami M, Amizuka N (2010) Immu- 83. Colditz J, Thiele S, Baschant U, Niehrs C, Bonewald LF, Hofbauer nolocalization of DMP1 and sclerostin in the epiphyseal trabecule LC, Rauner M (2018) Postnatal skeletal deletion of Dickkopf-1 and diaphyseal cortical bone of osteoprotegerin defcient mice. increases bone formation and bone volume in male and female Biomed Res 31:307–318 mice, despite increased sclerostin expression. J Bone Miner Res. 77. Brown JP, Prince RL, Deal C, Recker RR, Kiel DP, de Gre- https​://doi.org/10.1002/jbmr.3463 gorio LH, Hadji P, Hofbauer LC, Alvaro-Gracia JM, Wang H, 84. Florio M, Gunasekaran K, Stolina M, Li X, Liu L et al (2016) A Austin M, Wagman RB, Newmark R, Libanati C, San Martin bispecifc antibody targeting sclerostin and DKK-1 promotes bone J, Bone HG (2009) Comparison of the efect of denosumab and mass accrual and fracture repair. Nat Commun 7:11505 alendronate on BMD and biochemical markers of bone turnover

1 3